Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 72
Filtrar
1.
J Steroid Biochem Mol Biol ; 216: 106041, 2022 02.
Artigo em Inglês | MEDLINE | ID: mdl-34864206

RESUMO

The hypoxic microenvironment of cryptorchidism is an important factor to induce the impairment of the structure and function of Sertoli cells and thus lead to spermatogenesis loss or tumorigenesis. Dihydrotestosterone (DHT), as a potent nonaromatizable 5α-reduced androgen, has both positive and negative effect on pathological fibrosis process. However, it is still unknown whether DHT can regulate hypoxia-induced fibrosis of Sertoli cells. Herein, in this study, we evaluate the DHT level, two 5α-reductase isoforms, 5α-red1 and 5α-red2, as well as HIF-1α expression pattern in canine cryptorchidism and contralateral normal testis. Results showed that the abdominal testes presented low DHT levels and 5α-red1 and 5α-red2 expression, while significantly higher HIF-1α expression and ECM production compared with the scrotum. Moreover, we established a hypoxia-induced fibrosis model in canine Sertoli cells induced by cobalt chloride (CoCl2), and found that DHT inhibited the fibrosis of Sertoli cells in a dose-dependent manner. Meanwhile, DHT interfered with the TGF-ß signaling by reducing the expression of TGF-ßRI and TGF-ßRII and inhibiting the expression and phosphorylation of Smad2 and Smad3, while flutamide (androgen receptor inhibitor) inhibited these effects of DHT. Furthermore, use of LY2109761 (TGF-ß receptor type I/II inhibitor) to interfere with the TGF-ß/Smad pathway showed a similar effect with DHT suppression of the fibrosis in Sertoli cells. Our research data demonstrated that cryptorchidism is located in a hypoxic and DHT deficiency microenvironment. Moreover, supplementing DHT can alleviate the fibrosis process of Sertoli cells caused by hypoxia, which is associated with AR regulating the inhibition of TGF-ß/Smad signaling.


Assuntos
Hipóxia Celular/fisiologia , Di-Hidrotestosterona/farmacologia , Células de Sertoli/efeitos dos fármacos , Animais , Antifibróticos/farmacologia , Hipóxia Celular/efeitos dos fármacos , Células Cultivadas , Cães , Fibrose/patologia , Fibrose/prevenção & controle , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Masculino , Escroto/efeitos dos fármacos , Escroto/metabolismo , Escroto/patologia , Células de Sertoli/metabolismo , Células de Sertoli/patologia , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/antagonistas & inibidores , Proteínas Smad/metabolismo , Testículo/efeitos dos fármacos , Testículo/metabolismo , Testículo/patologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Fator de Crescimento Transformador beta/metabolismo
2.
Bioorg Med Chem ; 49: 116438, 2021 11 01.
Artigo em Inglês | MEDLINE | ID: mdl-34610571

RESUMO

Liver fibrosis is one of the most common pathological consequences of chronic liver diseases (CLD). To develop effective antifibrotic strategies, a novel class of 1-(substituted phenyl)-1,8-naphthalidine-3-carboxamide derivatives were designed and synthesized. By means of the collagen type I α 1 (COL1A1)-based screening and cytotoxicity assay in human hepatic stellate cell (HSC) line LX-2, seven compounds were screened out from total 60 derivatives with high inhibitory effect and relatively low cytotoxicity for further COL1A1 mRNA expression analysis. It was found that compound 17f and 19g dose-dependently inhibited the expression of fibrogenic markers, including α-smooth muscle actin (α-SMA), matrix metalloprotein 2 (MMP-2), connective tissue growth factor (CTGF) and transforming growth factor ß1 (TGFß1) on both mRNA and protein levels. Further mechanism studies indicated that they might suppress the hepatic fibrogenesis via inhibiting both PI3K/AKT/Smad and non-Smad JAK2/STAT3 signaling pathways. Furthermore, 19g administration attenuated hepatic histopathological injury and collagen accumulation, and reduced fibrogenesis-associated protein expression in liver tissues of bile duct ligation (BDL) rats, showing significant antifibrotic effect in vivo. These findings identified 1,8-naphthalidine derivatives as potent anti-hepatic fibrosis agents, and provided valuable information for further structure optimization.


Assuntos
1-Naftilamina/farmacologia , Descoberta de Drogas , Cirrose Hepática/tratamento farmacológico , 1-Naftilamina/síntese química , 1-Naftilamina/química , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Relação Dose-Resposta a Droga , Humanos , Janus Quinase 2/antagonistas & inibidores , Janus Quinase 2/metabolismo , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Estrutura Molecular , Fosfatidilinositol 3-Quinases/metabolismo , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Fator de Transcrição STAT3/antagonistas & inibidores , Fator de Transcrição STAT3/metabolismo , Proteínas Smad/antagonistas & inibidores , Proteínas Smad/metabolismo , Relação Estrutura-Atividade
3.
Biomed Pharmacother ; 141: 111806, 2021 Sep.
Artigo em Inglês | MEDLINE | ID: mdl-34246190

RESUMO

Diabetic nephropathy (DN) is a common disease, and patients often do not have satisfactory treatments. We investigated therapeutic effects of Fuxin Granules(FX) on DN and potential molecular mechanisms. We orally administered doses of FX to db/db mice for 10 weeks and measured total cholesterol (TC), triglycerides (TG), low-density lipoprotein cholesterol (LDL-C), and high-density lipoprotein cholesterol. H&E, PAS, Masson, and Oil Red O staining were used to observe the structure of kidneys and calculate indices of kidney function. We used pharmacological analysis to investigate potential mechanisms of FX. Relative mRNA and protein levels in the TGF-ß1/Smad, TGF-ß1/Smad, and VEGF/VEGFR2 pathways were examined. TC, TG, and LDL-C were markedly reduced, lipid accumulation was low, fibrosis reduced, kidney atrophy improved, kidney lipid droplet number significantly reduced, and glomerular filtration function improved by FX treatment. Multi-channel therapeutic effects in DN through the TGF-ß1/Smad and VEGF/VEGFR2 signaling pathways occurred, and FX substantially reduced expression of TGF-ß1 in the glomeruli. FX significantly inhibited TGF-ß1, Smad2/3 total protein levels, Smad2/3 phosphorylation mRNA levels of TGF-ß1, Smad2, and Smad3. eNOS, VEGFA, and VEGFR2 expression was regulated, levels of VEGFA and VEGFR2 were decreased, and FX increased eNOS. FX ameliorated symptoms of DN, resulting in marked improvement in hyperglycemia and hyperlipidemia and optimized structure and function of kidneys in db/db mice. FX efficacy was associated with the TGF-ß1/Smad and VEGF/VEGFR2 signaling pathways. We verified this potential mechanism and hope that this study will provide benefits for the clinical treatment of DN.


Assuntos
Nefropatias Diabéticas/metabolismo , Medicamentos de Ervas Chinesas/uso terapêutico , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Fator A de Crescimento do Endotélio Vascular/metabolismo , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/metabolismo , Animais , Nefropatias Diabéticas/tratamento farmacológico , Relação Dose-Resposta a Droga , Medicamentos de Ervas Chinesas/farmacologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Camundongos Transgênicos , Farmacologia em Rede/métodos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/fisiologia , Proteínas Smad/antagonistas & inibidores , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Fator A de Crescimento do Endotélio Vascular/antagonistas & inibidores , Receptor 2 de Fatores de Crescimento do Endotélio Vascular/antagonistas & inibidores
4.
Drug Des Devel Ther ; 15: 871-887, 2021.
Artigo em Inglês | MEDLINE | ID: mdl-33664566

RESUMO

PURPOSE: Taxifolin is a kind of dihydroflavone and is usually used as a food additive and health food for its antioxidant, anti-inflammatory, and anti-tumor activities. The purpose of this research is to probe into the hepatoprotective activity and the molecular mechanism of taxifolin. MATERIALS AND METHODS: The liver fibrosis model was established by intraperitoneal injection of 5 mL/kg body weight of CCl4 (20% CCl4 peanut oil solution), and taxifolin was dissolved with 0.9% physiological saline and administered intragastrically to mice. RESULTS: The results indicated that CCl4-induced significantly increased the serum alanine aminotransferase (ALT) and aspartate aminotransferase (AST) in mice. Histopathological examination showed severe hepatocyte necrosis and hepatic tissue lesion. Immunohistochemical staining and rt-PCR analysis demonstrated that the expressions of inducible nitric oxide synthetase (iNOS), cyclooxygenase-2 (COX-2), IL-1ß, IL-6, and TNF-α were increased. These changes were significantly reversed when treated with taxifolin. In addition, TUNEL staining and Bcl-2/Bax pathway confirmed that taxifolin significantly inhibited hepatocyte apoptosis. Besides, the research confirmed that taxifolin also inhibited the activation of hepatic stellate cells and the production of extracellular matrix (ECM) by regulating PI3K/AKT/mTOR and TGF-ß1/Smads pathways. CONCLUSION: Taxifolin inhibited inflammation, and attenuated CCl4-induced oxidative stress and cell apoptosis by regulating PI3K/AKT/mTOR and TGF-ß1/Smads pathways, which might in part contributed to taxifolin anti-hepatic fibrosis, further demonstrating that taxifolin may be an efficient hepatoprotective agent.


Assuntos
Anti-Inflamatórios não Esteroides/farmacologia , Tetracloreto de Carbono/antagonistas & inibidores , Larix/química , Cirrose Hepática/tratamento farmacológico , Quercetina/análogos & derivados , Animais , Anti-Inflamatórios não Esteroides/química , Anti-Inflamatórios não Esteroides/isolamento & purificação , Apoptose/efeitos dos fármacos , Relação Dose-Resposta a Droga , Hepatócitos/efeitos dos fármacos , Inflamação/tratamento farmacológico , Inflamação/metabolismo , Inflamação/patologia , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/patologia , Masculino , Camundongos , Camundongos Endogâmicos ICR , Conformação Molecular , Estresse Oxidativo/efeitos dos fármacos , Fosfatidilinositol 3-Quinases/metabolismo , Raízes de Plantas/química , Proteínas Proto-Oncogênicas c-akt/antagonistas & inibidores , Proteínas Proto-Oncogênicas c-akt/metabolismo , Quercetina/química , Quercetina/isolamento & purificação , Quercetina/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/antagonistas & inibidores , Proteínas Smad/metabolismo , Relação Estrutura-Atividade , Serina-Treonina Quinases TOR/antagonistas & inibidores , Serina-Treonina Quinases TOR/metabolismo , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Fator de Crescimento Transformador beta1/metabolismo
5.
J Ethnopharmacol ; 270: 113838, 2021 Apr 24.
Artigo em Inglês | MEDLINE | ID: mdl-33460756

RESUMO

ETHNOPHARMACOLOGICAL RELEVANCE: Myocardial fibrosis after myocardial infarction (MI) leads to cardiac remodeling and loss of function. Taohong siwu decoction (THSWD), a well-known traditional Chinese medicinal prescription, has been clinically used to treat various cardiovascular and cerebrovascular diseases, but its potential functions in myocardial fibrosis after MI remain uncharacterized. AIM OF THE STUDY: The purpose of current study was to explore the potential mechanism action and anti-myocardial fibrosis effects of treatment with THSWD in vivo and in vitro. MATERIALS AND METHODS: Mouse underwent ligation of coronary artery to induce MI and divided equally into the sham group, model group and THSWD treatment groups. After 4 weeks, the effects of THSWD treatment on cardiac function were estimated by echocardiography. HE staining was used to detect the pathologic changes and Masson trichrome staining was used to estimate tissue fibrosis. To further explore the regulatory molecular mechanisms of THSWD, transcriptome analysis was performed. Furthermore, in vitro, we investigated the effect of THSWD on cell proliferation and collagen deposition in primary cardiac fibrosis cells and its possible mechanism of action. Overexpression of TGFBR1 was achieved by infection with an adenovirus vector encoding TGFBR1. RESULTS: Treatment with THSWD significantly decreased myocardial fibrosis and recovered cardiac function in the post-MI mouse. The transcriptomics data imply that the TGF-ß pathway might be a target in the anti-fibrosis effect of THSWD. THSWD inhibits TGF-ß1-induced proliferation of primary cardiac fibroblasts. THSWD decreased collagen expression and TGFBR1 and Smad2/3 phosphorylation. Moreover, the inhibitory effect of THSWD on CFs proliferation and collagen deposition, as well as TGFBR1 signaling pathway-associated proteins expression was partially abrogated by overexpression of TGFBR1. CONCLUSION: Collectively, the results implicate that THSWD attenuates myocardial fibrosis by inhibiting fibrosis proliferation and collagen deposition via inhibiting TGFBR1, and might be a potential therapeutic agent for treatment of myocardial fibrosis post-MI.


Assuntos
Colágeno/metabolismo , Medicamentos de Ervas Chinesas/farmacologia , Medicamentos de Ervas Chinesas/uso terapêutico , Fibrose/tratamento farmacológico , Receptor do Fator de Crescimento Transformador beta Tipo I/metabolismo , Transdução de Sinais/efeitos dos fármacos , Animais , Proliferação de Células/efeitos dos fármacos , Colágeno/antagonistas & inibidores , Modelos Animais de Doenças , Medicamentos de Ervas Chinesas/química , Fibroblastos/efeitos dos fármacos , Fibroblastos/metabolismo , Fibrose/etiologia , Fibrose/metabolismo , Fibrose/patologia , Masculino , Camundongos Endogâmicos C57BL , Infarto do Miocárdio/complicações , Infarto do Miocárdio/diagnóstico por imagem , Miocárdio/metabolismo , Miocárdio/patologia , Cultura Primária de Células , Ratos Sprague-Dawley , Receptor do Fator de Crescimento Transformador beta Tipo I/antagonistas & inibidores , Receptor do Fator de Crescimento Transformador beta Tipo I/genética , Proteínas Smad/antagonistas & inibidores , Proteínas Smad/metabolismo , Transcriptoma/efeitos dos fármacos
6.
Pediatr Res ; 89(3): 502-509, 2021 02.
Artigo em Inglês | MEDLINE | ID: mdl-32365352

RESUMO

BACKGROUND: Bronchopulmonary dysplasia remains one of the most common complications of prematurity, despite significant improvements in perinatal care. Functional modeling of human lung development and disease, like BPD, is limited by our ability to access the lung and to maintain relevant progenitor cell populations in culture. METHODS: We supplemented Rho/SMAD signaling inhibition with mTOR inhibition to generate epithelial basal cell-like cell lines from tracheal aspirates of neonates. RESULTS: Single-cell RNA-sequencing confirmed the presence of epithelial cells in tracheal aspirates obtained from intubated neonates. Using Rho/SMAD/mTOR triple signaling inhibition, neonatal tracheal aspirate-derived (nTAD) basal cell-like cells can be expanded long term and retain the ability to differentiate into pseudostratified airway epithelium. CONCLUSIONS: Our data demonstrate that neonatal tracheal aspirate-derived epithelial cells can provide a novel ex vivo human cellular model to study neonatal lung development and disease. IMPACT: Airway epithelial basal cell-like cell lines were derived from human neonatal tracheal aspirates. mTOR inhibition significantly extends in vitro proliferation of neonatal tracheal aspirate-derived basal cell-like cells (nTAD BCCs). nTAD BCCs can be differentiated into functional airway epithelium. nTAD BCCs provide a novel model to investigate perinatal lung development and diseases.


Assuntos
Células Epiteliais/efeitos dos fármacos , Proteínas Smad/antagonistas & inibidores , Serina-Treonina Quinases TOR/antagonistas & inibidores , Traqueia/citologia , Quinases Associadas a rho/antagonistas & inibidores , Sequência de Bases , Líquidos Corporais/citologia , Displasia Broncopulmonar , Diferenciação Celular/efeitos dos fármacos , Divisão Celular/efeitos dos fármacos , Células Cultivadas , Células Epiteliais/química , Células Epiteliais/citologia , Humanos , Recém-Nascido , Cultura Primária de Células , Análise de Célula Única , Sirolimo/farmacologia , Proteínas Smad/fisiologia , Células-Tronco/citologia , Células-Tronco/efeitos dos fármacos , Sucção , Serina-Treonina Quinases TOR/fisiologia , Quinases Associadas a rho/fisiologia
7.
Eur J Gastroenterol Hepatol ; 32(6): 669-677, 2020 06.
Artigo em Inglês | MEDLINE | ID: mdl-32282548

RESUMO

In the 'treat-to-target' era of inflammatory bowel disease (IBD) management, small molecule drugs (SMDs) represent a promising alternative to biomolecular drugs. Moreover, increasing failure rates of anti-tumor necrosis factor α agents have contributed to the development of new molecules with different mechanisms of action and bioavailability. This review focuses on the positioning of new, orally targeted therapies in the treatment algorithm of both Crohn's disease (CD) and ulcerative colitis (UC), with special consideration to their efficacy and safety. We performed a comprehensive search of PubMed and clinical trial registries to identify randomized controlled trials assessing SMDs in adult patients with moderate-to-severe IBD, irrespective of previous exposure to other biologics. In this review, we included 15 double-blind, placebo-controlled trials that assessed the efficacy and safety of Janus kinase inhibitors, sphingosine-1-phosphate modulators (S1P), SMAD blockers, phosphodiesterase 4 inhibitors and α-4 antagonists. The primary endpoints in UC were achieved for tofacitinib in the phase III OCTAVE study and AJM-300, with a favorable safety profile. S1P receptor agonists, such as etrasimod and ozanimod, demonstrated favorable results in induction studies. For CD, filgotinib and upadacitinib also met the primary outcome criteria. Available data have demonstrated so far that SMDs have an advantageous safety and efficacy profile. However, their use in a clinical setting will eventually require a personalized, mechanism-based therapeutic approach.


Assuntos
Doenças Inflamatórias Intestinais/tratamento farmacológico , Bibliotecas de Moléculas Pequenas/uso terapêutico , Humanos , Integrina alfa4/antagonistas & inibidores , Inibidores de Janus Quinases/uso terapêutico , Inibidores da Fosfodiesterase 4/uso terapêutico , Proteínas Smad/antagonistas & inibidores , Moduladores do Receptor de Esfingosina 1 Fosfato/uso terapêutico
8.
Acta Pharmacol Sin ; 41(5): 670-677, 2020 May.
Artigo em Inglês | MEDLINE | ID: mdl-31804606

RESUMO

Renal fibrosis is considered as the pathway of almost all kinds of chronic kidney diseases (CKD) to the end stage of renal diseases (ESRD). Ganoderic acid (GA) is a group of lanostane triterpenes isolated from Ganoderma lucidum, which has shown a variety of pharmacological activities. In this study we investigated whether GA exerted antirenal fibrosis effect in a unilateral ureteral obstruction (UUO) mouse model. After UUO surgery, the mice were treated with GA (3.125, 12.5, and 50 mg· kg-1 ·d-1, ip) for 7 or 14 days. Then the mice were sacrificed for collecting blood and kidneys. We showed that GA treatment dose-dependently attenuated UUO-induced tubular injury and renal fibrosis; GA (50 mg· kg-1 ·d-1) significantly ameliorated renal disfunction during fibrosis progression. We further revealed that GA treatment inhibited the extracellular matrix (ECM) deposition in the kidney by suppressing the expression of fibronectin, mainly through hindering the over activation of TGF-ß/Smad signaling. On the other hand, GA treatment significantly decreased the expression of mesenchymal cell markers alpha-smooth muscle actin (α-SMA) and vimentin, and upregulated E-cadherin expression in the kidney, suggesting the suppression of tubular epithelial-mesenchymal transition (EMT) partially via inhibiting both TGF-ß/Smad and MAPK (ERK, JNK, p38) signaling pathways. The inhibitory effects of GA on TGF-ß/Smad and MAPK signaling pathways were confirmed in TGF-ß1-stimulated HK-2 cell model. GA-A, a GA monomer, was identified as a potent inhibitor on renal fibrosis in vitro. These data demonstrate that GA or GA-A might be developed as a potential therapeutic agent in the treatment of renal fibrosis.


Assuntos
Proteínas Smad/antagonistas & inibidores , Fator de Crescimento Transformador beta/antagonistas & inibidores , Triterpenos/farmacologia , Obstrução Ureteral/tratamento farmacológico , Animais , Células Cultivadas , Modelos Animais de Doenças , Relação Dose-Resposta a Droga , Humanos , Injeções Intraperitoneais , Sistema de Sinalização das MAP Quinases/efeitos dos fármacos , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Triterpenos/administração & dosagem , Obstrução Ureteral/metabolismo , Obstrução Ureteral/cirurgia
9.
J Cell Physiol ; 235(1): 429-441, 2020 01.
Artigo em Inglês | MEDLINE | ID: mdl-31230364

RESUMO

Colorectal cancer (CRC) is still a fatal health problem around the world. The underlying mechanisms of CRC have not been fully elucidated. N-myc interactor (NMI) acts as an oncogene or a tumor-suppressor gene in several kinds of cancers but CRC. Here, the expression of NMI was found higher in CRC tissues and cells. Higher expression of NMI indicated the poorer prognosis of CRC patients. Moreover, the proliferation of CRC cells was suppressed significantly after we silenced the expression of NMI, while overexpression of NMI promoted CRC cell proliferation. Flow cytometry demonstrated that NMI promoted cell proliferation through facilitating cell transition from the G1 phase to the S phase. Furthermore, it was found that NMI suppressed the phosphorylation of Smad3 by upregulating the expression of STAT1. The effect of NMI depletion on cell proliferation could be reversed by using Smad3 inhibitor SIS3. In summary, our findings demonstrated that NMI promoted cell proliferation via TGFß/Smad pathway and could indicate the prognosis of patients with CRC.


Assuntos
Neoplasias Colorretais/patologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Fator de Transcrição STAT1/metabolismo , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/metabolismo , Apoptose/fisiologia , Linhagem Celular Tumoral , Proliferação de Células/fisiologia , Transformação Celular Neoplásica/patologia , Neoplasias Colorretais/genética , Humanos , Peptídeos e Proteínas de Sinalização Intracelular/genética , Isoquinolinas/farmacologia , Prognóstico , Piridinas/farmacologia , Pirróis/farmacologia , Interferência de RNA , RNA Interferente Pequeno/genética , Fator de Transcrição STAT1/genética , Proteínas Smad/antagonistas & inibidores , Ativação Transcricional/genética
10.
Int J Biochem Cell Biol ; 116: 105613, 2019 11.
Artigo em Inglês | MEDLINE | ID: mdl-31539631

RESUMO

Renal tubular epithelial-mesenchymal transition (EMT) is the main pathological change in diabetic renal tubulointerstitial fibrosis. Mounting evidence indicates that the inhibitor of differentiation 2 (Id2) protein acts as a negative regulatory factor in organ fibrosis and can inhibit or reverse the process of fibrosis. However, its specific regulatory mechanism is not clear. Diabetes mellitus (DM) rat models were established by injecting rats with streptozotocin and sacrificing them after 16 weeks. Rat renal tubular epithelial cells (NRK-52E) were cultured with normal and high glucose. Immunohistochemical analysis, double immunofluorescence staining, co-immunoprecipitation, Western blot analysis, and real-time polymerase chain reaction were used to determine the expression of Id2, Twist, Smad1/5/8, E-cadherin, α-smooth muscle actin (α-SMA), and collagen Ⅳ. The results showed that bone morphogenetic protein-7 (BMP-7) upregulated the expression of Id2 against high-glucose-induced EMT and extracellular matrix secretion. Moreover, only the simultaneous knockdown of Smad1, Smad5, and Smad8 downregulated the expression of Id2, which was not altered by the individual knockdown of Smad1, Smad5, and Smad8. Basic helix-loop-helix (bHLH) transcription factors were essential for Id2 to regulate the role of downstream target genes, and Twist was a bHLH transcription factor. Therefore, the expression of Twist was examined in this study. Twist was found to be highly expressed in the kidney of DM rats and renal tubular epithelial cells cultured with high glucose. The overexpression of Id2 did not alter the expression of Twist, but the interaction between Id2 and Twist was enhanced. In conclusion, the data showed the specific mechanism underlying Id2 negative regulation in diabetic renal tubulointerstitial fibrosis.


Assuntos
Proteína Morfogenética Óssea 7/genética , Diabetes Mellitus Experimental/genética , Nefropatias Diabéticas/genética , Proteína 2 Inibidora de Diferenciação/genética , Nefrite Intersticial/genética , Proteínas Smad/genética , Proteína 1 Relacionada a Twist/genética , Actinas/genética , Actinas/metabolismo , Animais , Proteína Morfogenética Óssea 7/metabolismo , Caderinas/genética , Caderinas/metabolismo , Linhagem Celular , Colágeno Tipo IV/genética , Colágeno Tipo IV/metabolismo , Diabetes Mellitus Experimental/induzido quimicamente , Diabetes Mellitus Experimental/metabolismo , Diabetes Mellitus Experimental/patologia , Nefropatias Diabéticas/induzido quimicamente , Nefropatias Diabéticas/metabolismo , Nefropatias Diabéticas/patologia , Células Epiteliais/efeitos dos fármacos , Células Epiteliais/metabolismo , Transição Epitelial-Mesenquimal/efeitos dos fármacos , Matriz Extracelular/química , Matriz Extracelular/efeitos dos fármacos , Regulação da Expressão Gênica , Glucose/farmacologia , Proteína 2 Inibidora de Diferenciação/metabolismo , Masculino , Nefrite Intersticial/induzido quimicamente , Nefrite Intersticial/metabolismo , Nefrite Intersticial/patologia , RNA Interferente Pequeno/genética , RNA Interferente Pequeno/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Proteínas Smad/antagonistas & inibidores , Proteínas Smad/metabolismo , Estreptozocina/administração & dosagem , Proteína 1 Relacionada a Twist/metabolismo
11.
Biochim Biophys Acta Mol Cell Res ; 1866(4): 588-597, 2019 04.
Artigo em Inglês | MEDLINE | ID: mdl-30615900

RESUMO

Increased GP73 expression in hepatocytes from patients with acute hepatitis, through disease progression to cirrhosis and chronic liver disease suggests that progressive tissue remodeling and fibrogenesis are driving forces for GP73 upregulation. Nevertheless, details about regulation of GP73 expression and its biological functions remain elusive and await further characterization. In this study, we demonstrate that GP73 is a direct target of TGF-ß1 transcriptional regulation. Its induced expression inhibits TGF-ß-Smad mediated growth suppression. On the other hand, elevated GP73 results in upregulation of ERK/Akt signaling induced by TGF-ß1. Mechanistically, upregulation of lipid raft and caveolae-1 induced by GP73 overexpression mediates its regulatory effect on TGF-ß1 signaling. Notably, lipid raft expression is elevated in HCC tumors and tissues with higher GP73 expression yield more intensive Flotillin staining. Our results establish the linkage between GP73 and TGF-ß signaling, indicating that GP73 may promote HCC tumorigenesis by selectively regulating TGF-ß signaling through lipid raft modulation.


Assuntos
Proteínas de Membrana/metabolismo , Transdução de Sinais , Proteínas Smad/antagonistas & inibidores , Fator de Crescimento Transformador beta1/fisiologia , Animais , Carcinoma Hepatocelular/metabolismo , Caveolina 1/metabolismo , Linhagem Celular , Células Cultivadas , MAP Quinases Reguladas por Sinal Extracelular/metabolismo , Regulação da Expressão Gênica , Humanos , Neoplasias Hepáticas/metabolismo , Masculino , Microdomínios da Membrana/metabolismo , Proteínas de Membrana/biossíntese , Proteínas de Membrana/genética , Camundongos , Camundongos Endogâmicos BALB C , Camundongos Nus , Fosfoproteínas/biossíntese , Fosfoproteínas/genética , Transcrição Gênica
12.
Cell Metab ; 29(3): 638-652.e5, 2019 03 05.
Artigo em Inglês | MEDLINE | ID: mdl-30581122

RESUMO

Small-molecule inhibitors of dual-specificity tyrosine-regulated kinase 1A (DYRK1A) induce human beta cells to proliferate, generating a labeling index of 1.5%-3%. Here, we demonstrate that combined pharmacologic inhibition of DYRK1A and transforming growth factor beta superfamily (TGFßSF)/SMAD signaling generates remarkable further synergistic increases in human beta cell proliferation (average labeling index, 5%-8%, and as high as 15%-18%), and increases in both mouse and human beta cell numbers. This synergy reflects activation of cyclins and cdks by DYRK1A inhibition, accompanied by simultaneous reductions in key cell-cycle inhibitors (CDKN1C and CDKN1A). The latter results from interference with the basal Trithorax- and SMAD-mediated transactivation of CDKN1C and CDKN1A. Notably, combined DYRK1A and TGFß inhibition allows preservation of beta cell differentiated function. These beneficial effects extend from normal human beta cells and stem cell-derived human beta cells to those from people with type 2 diabetes, and occur both in vitro and in vivo.


Assuntos
Diabetes Mellitus Tipo 2 , Harmina/farmacologia , Células Secretoras de Insulina , Inibidores da Monoaminoxidase/farmacologia , Fator de Crescimento Transformador beta/antagonistas & inibidores , Adolescente , Adulto , Idoso , Animais , Linhagem Celular , Proliferação de Células , Diabetes Mellitus Tipo 2/tratamento farmacológico , Diabetes Mellitus Tipo 2/metabolismo , Feminino , Histona-Lisina N-Metiltransferase/antagonistas & inibidores , Humanos , Células Secretoras de Insulina/efeitos dos fármacos , Células Secretoras de Insulina/patologia , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Pessoa de Meia-Idade , Proteína de Leucina Linfoide-Mieloide/antagonistas & inibidores , Proteínas Serina-Treonina Quinases/antagonistas & inibidores , Proteínas Tirosina Quinases/antagonistas & inibidores , Proteínas Smad/antagonistas & inibidores , Células-Tronco , Adulto Jovem , Quinases Dyrk
13.
Biochem Biophys Res Commun ; 504(1): 46-53, 2018 09 26.
Artigo em Inglês | MEDLINE | ID: mdl-30172378

RESUMO

BACKGROUND: To investigate the effects and underlying molecular mechanisms of FoxG1 expression on glioblastoma multiforme (GBM) models. METHODS: Expression levels of FoxG1 and other cancer-related biomarkers were evaluated by qRT-PCR, immunoblotting and immunohistochemistry. Crystal violet staining and MTT assay and were applied in this study to verify cell proliferation ability and viability of GBM cell models with/without drug treatment. RESULTS: Immunohistochemical and qRT-PCR assays showed that endogenous FoxG1 expression levels were positively correlated to the GBM disease progression. Overexpression of FoxG1 protein resulted in increased cell viability, G2/M cell cycle arrest, as well as the downregulation of p21 and cyclin B1. In addition, western blot assays reported that enforced expression of FoxG1 suppressed GAPF and facilitated the expression of Sox2 and Sox5. Meanwhile the downstream targets of FoxG1, such as FoxO1 and pSmad1/5/8 were activated. Overexpression of FoxG1 under TMZ treatment restored the cell viability as well as the expression levels of Sox2 and Sox5, yet downregulated expression levels of p21 and cyclin B1. The downstream FoxG1-induced FoxO/Smad signaling was re-inhibited under TMZ treatments. CONCLUSIONS: Our findings suggest that FoxG1 functions as an onco-factor by promoting proliferation, as well as inhibiting differential responses in glioblastoma by downregulating FoxO/Smad signaling.


Assuntos
Neoplasias Encefálicas/metabolismo , Fatores de Transcrição Forkhead/metabolismo , Glioblastoma/metabolismo , Proteínas do Tecido Nervoso/metabolismo , Antineoplásicos Alquilantes/farmacologia , Neoplasias Encefálicas/patologia , Diferenciação Celular , Linhagem Celular Tumoral , Proliferação de Células , Progressão da Doença , Regulação para Baixo , Proteína Forkhead Box O1/antagonistas & inibidores , Fatores de Transcrição Forkhead/genética , Glioblastoma/patologia , Humanos , Proteínas do Tecido Nervoso/genética , Transdução de Sinais , Proteínas Smad/antagonistas & inibidores , Temozolomida/farmacologia
14.
Cell Physiol Biochem ; 49(1): 160-171, 2018.
Artigo em Inglês | MEDLINE | ID: mdl-30134228

RESUMO

BACKGROUND/AIMS: Lymphangiogenesis plays an important role in the pathogenesis of inflammatory bowel diseases (IBD), and vascular endothelial growth factor-C (VEGF-C) is a powerful lymphangiogenic factor. Adipose-derived stem cells (ADSCs) are a promising therapeutic modality for several diseases because ADSCs secret growth factors and exosomes, which modulate hostile microenvironments affected by diseases. However, the effect of exosomes on VEGF-C-dependent lymphangiogenesis and its mechanism remain unclear. METHODS: ADSCs were cultured in media with or without recombinant VEGF-C and exosomes were extracted from conditioned medium (CM). Lymphatic endothelial cells (LECs) were treated with ADSCs-derived exosomes, then proliferation, migration and tube formation of LECs were assayed using cell counting Kit-8 (CCK-8), transwell chamber inserts and matrigel-based tube formation assay respectively. RESULTS: We identified significantly higher levels of miR-132 in exosomes isolated from VEGF-C-treated ADSCs (ADSCs/VEGF-C) than in those from ADSCs control. miR-132 was directly transferred from ADSCs to the LECs by the mediation of exosomes. The exosomes from ADSCs/VEGF-C promoted LECs proliferation, migration, and tube formation more potently than the exosomes from ADSCs, whereas pretreatment of ADSCs with miR-132 inhibitor attenuates VEGF-C-dependent lymphangiogenic response. Finally we reveal that miR-132 promotes lymphangiogenic response by directly targeting Smad-7 and regulating TGF-ß/Smad signaling. CONCLUSION: These data provide new insights into the role of ADSCs-derived exosomes as an important player in VEGF-C-dependent lymphangiogenesis.


Assuntos
Exossomos/metabolismo , Linfangiogênese/efeitos dos fármacos , Transdução de Sinais/efeitos dos fármacos , Fator C de Crescimento do Endotélio Vascular/farmacologia , Regiões 3' não Traduzidas , Tecido Adiposo/citologia , Proliferação de Células/efeitos dos fármacos , Células Cultivadas , Meios de Cultivo Condicionados/farmacologia , Proteínas de Choque Térmico HSP70/metabolismo , Humanos , MicroRNAs/genética , MicroRNAs/metabolismo , Neovascularização Fisiológica/efeitos dos fármacos , Proteínas Smad/antagonistas & inibidores , Proteínas Smad/genética , Proteínas Smad/metabolismo , Células-Tronco/citologia , Células-Tronco/metabolismo , Tetraspanina 30/metabolismo , Fator de Crescimento Transformador beta/metabolismo
15.
Endocrinology ; 159(5): 2062-2074, 2018 05 01.
Artigo em Inglês | MEDLINE | ID: mdl-29579175

RESUMO

The luteinizing hormone receptor (LHCGR) is expressed at low levels in mural granulosa cells and cumulus cells of antral follicles and is induced dramatically in granulosa cells but not in cumulus cells by follicle-stimulating hormone (FSH). Therefore, we hypothesized that FSH not only activates transcription factors controlling Lhcgr expression but also alters other events to permit and enhance Lhcgr expression in granulosa cells but not in cumulus cells. In granulosa cells, the level of DNA methylation in the Lhcgr promoter region was significantly decreased by equine chorionic gonadotropin (eCG) in vivo. However, in cumulus cells, hypermethylation of the Lhcgr promoter remained after eCG stimulation. eCG induced estrogen production from testosterone (T) and retinoic acid (RA) synthesis in granulosa cells. When either T or RA in the presence or absence of FSH was added to granulosa cell cultures, the combined treatment with FSH and RA induced demethylation of Lhcgr-promoter region and Lhcgr expression. FSH-dependent RA synthesis was negatively regulated by coculture of granulosa cells with denuded oocytes, suggesting that oocyte-secreted factors downregulate RA production in cumulus cells where Lhcgr expression was not induced. Strikingly, treatment of cultured cumulus-oocyte complexes with a SMAD inhibitor, SB431542, significantly induced RA production, demethylation of Lhcgr-promoter region, and Lhcgr expression in cumulus cells. These results indicate the demethylation of the Lhcgr-promoter region is mediated, at least in part, by RA synthesis and is a key mechanism regulating the cell type-specific differentiation during follicular development.


Assuntos
Células do Cúmulo/metabolismo , Desmetilação do DNA , Metilação de DNA/genética , Hormônio Foliculoestimulante/metabolismo , Células da Granulosa/metabolismo , Receptores do LH/genética , Animais , Benzamidas/farmacologia , Células Cultivadas , Gonadotropina Coriônica/farmacologia , Células do Cúmulo/efeitos dos fármacos , Desmetilação do DNA/efeitos dos fármacos , Metilação de DNA/efeitos dos fármacos , Dioxóis/farmacologia , Estrogênios/metabolismo , Feminino , Hormônio Foliculoestimulante/farmacologia , Regulação da Expressão Gênica , Células da Granulosa/efeitos dos fármacos , Hormônios/farmacologia , Camundongos , Ovário , Regiões Promotoras Genéticas , Substâncias para o Controle da Reprodução/farmacologia , Proteínas Smad/antagonistas & inibidores , Testosterona/metabolismo , Testosterona/farmacologia , Tretinoína/metabolismo , Tretinoína/farmacologia
16.
Arthritis Res Ther ; 20(1): 46, 2018 03 15.
Artigo em Inglês | MEDLINE | ID: mdl-29544542

RESUMO

BACKGROUND: Transforming growth factor-ß (TGF-ß)/Smad signaling is well known to play a critical role in the pathogenesis of systemic sclerosis (SSc). We previously developed an artificial molecule, the histidine-pyridine-histidine ligand derivative HPH-15, which may have an antifibrotic effect. The purpose of the present study was to clarify the effects of this drug in human skin fibroblasts and in a preclinical model of SSc. METHODS: The effects of HPH-15 on expression of extracellular matrix components and TGF-ß signaling in human dermal fibroblasts were analyzed. The antifibrotic properties of HPH-15 and its mechanisms were also examined in a bleomycin-induced skin fibrosis mouse model. RESULTS: HPH-15 suppressed the TGF-ß-induced phosphorylation of Smad3 and inhibited the expression of collagen I, fibronectin 1, connective tissue growth factor, and α-smooth muscle actin induced by TGF-ß in cultured human skin fibroblasts. In the bleomycin-induced skin fibrosis model, oral administration of HPH-15 protected against the development of skin fibrosis and ameliorated established skin fibrosis. Additionally, HPH-15 suppressed the phosphorylation of Smad3 in various cells, including macrophages in the bleomycin-injected skin. Further, in the treated mice, dermal infiltration of proinflammatory macrophages (CD11b+Ly6Chi) and M2 profibrotic macrophages (CD11b+CD204+ or CD11b+CD206+) was significantly decreased during the early and late stages, respectively. HPH-15 treatment resulted in decreased messenger RNA (mRNA) expression of the M2 macrophage markers arginase 1 and Ym-1 in the skin, whereas it inversely augmented expression of Friend leukemia integration 1 and Krüppel-like factor 5 mRNAs, the transcription factors that repress collagen synthesis. No apparent adverse effects of HPH-15 were found during the treatment. CONCLUSIONS: HPH-15 may inhibit skin fibrosis by inhibiting the phosphorylation of Smad3 in dermal fibroblasts and possibly in macrophages. Our results demonstrate several positive qualities of HPH-15, including oral bioavailability, a good safety profile, and therapeutic effectiveness. Thus, this TGF-ß/Smad inhibitor is a potential candidate therapeutic for SSc clinical trials.


Assuntos
Histidina/farmacologia , Piridinas/farmacologia , Transdução de Sinais/efeitos dos fármacos , Pele/efeitos dos fármacos , Proteínas Smad/antagonistas & inibidores , Fator de Crescimento Transformador beta/antagonistas & inibidores , Animais , Células Cultivadas , Fibroblastos/efeitos dos fármacos , Fibroblastos/patologia , Fibrose/tratamento farmacológico , Fibrose/patologia , Histidina/química , Histidina/uso terapêutico , Humanos , Recém-Nascido , Ligantes , Masculino , Camundongos , Camundongos Endogâmicos C57BL , Piridinas/química , Piridinas/uso terapêutico , Transdução de Sinais/fisiologia , Pele/patologia , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo
17.
Int J Mol Med ; 41(1): 51-60, 2018 Jan.
Artigo em Inglês | MEDLINE | ID: mdl-29115380

RESUMO

The present study aimed to assess the effects of therapy with adiponectin (APN) gene-modified adipose-derived stem cells (ADSCs) on pulmonary arterial hypertension (PAH) in rats and the underlying cellular and molecular mechanisms. ADSCs were successfully isolated from the rats and characterized. ADSCs were effectively infected with the green fluorescent protein (GFP)-empty (ADSCs-V) or the APN-GFP (ADSCs-APN) lentivirus and the APN expression was evaluated by ELISA. Sprague-Dawley rats were administered monocrotaline (MCT) to develop PAH. The rats were treated with MCT, ADSCs, ADSCs-V and ADSCs-APN. Then ADSCs-APN in the lung were investigated by confocal laser scanning microscopy and western blot analysis. Engrafted ADSCs in the lung were located around the vessels. Mean pulmonary arterial pressure (mPAP) and the right ventricular hypertrophy index (RVHI) in the ADSCs-APN-treated mice were significantly decreased as compared with the ADSCs and ADSCs-V treatments. Pulmonary vascular remodeling was assessed. Right ventricular (RV) function was evaluated by echocardiography. We found that pulmonary vascular remodeling and the parameters of RV function were extensively improved after ADSCs-APN treatment when compared with ADSCs and ADSCs-V treatment. Pulmonary artery smooth muscle cells (PASMCs) were isolated from the PAH rats. The antiproliferative effect of APN on PASMCs was assayed by Cell Counting Kit-8. The influence of APN and specific inhibitors on the levels of bone morphogenetic protein (BMP), adenosine monophosphate activated protein kinase (AMPK), and small mothers against decapentaplegia (Smad) pathways was detected by western blot analysis. We found that APN suppressed the proliferation of PASMCs isolated from the PAH rats by regulating the AMPK/BMP/Smad pathway. This effect was weakened by addition of the AMPK inhibitor (compound C) and BMP2 inhibitor (noggin). Therefore, combination treatment with ADSCs and APN effectively attenuated PAH in rats by inhibiting PASMC proliferation and regulating the AMPK/BMP/Smad pathway.


Assuntos
Adiponectina/administração & dosagem , Proliferação de Células/genética , Hipertensão Pulmonar/terapia , Miócitos de Músculo Liso/efeitos dos fármacos , Quinases Proteína-Quinases Ativadas por AMP , Adipócitos/transplante , Adiponectina/genética , Tecido Adiposo/citologia , Tecido Adiposo/transplante , Animais , Proteína Morfogenética Óssea 2/antagonistas & inibidores , Proteína Morfogenética Óssea 2/genética , Proliferação de Células/efeitos dos fármacos , Terapia Genética , Humanos , Hipertensão Pulmonar/genética , Hipertensão Pulmonar/patologia , Lentivirus/genética , Miócitos de Músculo Liso/metabolismo , Proteínas Quinases/efeitos dos fármacos , Proteínas Quinases/genética , Artéria Pulmonar/efeitos dos fármacos , Artéria Pulmonar/patologia , Ratos , Transdução de Sinais/efeitos dos fármacos , Transdução de Sinais/genética , Proteínas Smad/antagonistas & inibidores , Proteínas Smad/genética
18.
Exp Cell Res ; 361(1): 192-198, 2017 12 01.
Artigo em Inglês | MEDLINE | ID: mdl-29069575

RESUMO

Pulmonary arterial hypertension (PAH), characterized by excessive proliferation and apoptosis resistance of pulmonary artery smooth muscle cells (PASMCs), is closely associated with the imbalance in vasoactive mediators and massive remodeling of pulmonary vasculature. DJ-1/park7, a multifunctional protein, plays a critical defense role in several cytobiological activity, such as transcriptional regulation, anti-oxidative stress and tumor formation. In this study, we investigated the effects of DJ-1 on hypoxia-induced PAH model rats and PASMCs, as well as its possible molecular mechanism. First, the low expressions of DJ-1 and caveolin-1 (Cav-1) were synchronously detected in lung tissue of PAH model rats and hypoxia-induced PASMCs by Western blot. Then, the DJ-1 wild type (WT) or Knock out (KO) rats were exposed to chronic hypoxia to mimic a hypoxic PAH condition. The protein level of Cav-1 was markedly decreased in the tissue of DJ-1 KO rats, and additionally lower in tissue of the hypoxia group than that in the normoxia group for DJ-1 WT and KO rats. In vivo, hemodynamic data showed that the pulmonary arterial pressure (mPAP), right ventricle systolic pressure (RVSP) and pulmonary arterial systolic pressure (PASP), as well as the weight of the right ventricle/left ventricle plus septum (RV/LV+S) ratio of PAH model rats were higher in the DJ-1 KO group than those in the DJ-1 WT group. Moreover, knockout of DJ-1 also results in the phenotype switch from contractile to synthetic PASMC, which is reflected by reduced calponin and SM22α expressions. In vitro, DJ-1 overexpression reversed hypoxia-induced elevation of PASMC cell proliferation, migration and Ca2+ concentration, which were not obviously observed in Cav-1 shRNA (sh-Cav-1) and DJ-1 co-transfected cells. Then the increased levels of calponin and SM22α were detected in the DJ-1 group; similarly those levels were not changed in the DJ-1+sh-Cav-1 group. Finally, the expression of TGFß1, p-Smad2 and p-Smad3 were obviously decreased in the ad-DJ-1 group, however those were all elevated in the DJ-1 and sh-Cav-1 co-transfected groups. In conclusion, these results indicate that DJ-1 may alleviate hypoxia-induced PASMCs injury by Cav-1 through inhibiting the TGFß/Smad signaling pathway.


Assuntos
Caveolina 1/metabolismo , Hipertensão Pulmonar/patologia , Músculo Liso Vascular/patologia , Proteína Desglicase DJ-1/metabolismo , Artéria Pulmonar/patologia , Proteínas Smad/antagonistas & inibidores , Fator de Crescimento Transformador beta1/antagonistas & inibidores , Animais , Animais Geneticamente Modificados , Apoptose , Caveolina 1/genética , Movimento Celular , Proliferação de Células , Células Cultivadas , Regulação da Expressão Gênica , Hipertensão Pulmonar/etiologia , Hipertensão Pulmonar/metabolismo , Hipóxia/fisiopatologia , Masculino , Músculo Liso Vascular/metabolismo , Proteína Desglicase DJ-1/genética , Artéria Pulmonar/metabolismo , Ratos , Ratos Sprague-Dawley , Transdução de Sinais , Proteínas Smad/genética , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta1/genética , Fator de Crescimento Transformador beta1/metabolismo
19.
J Recept Signal Transduct Res ; 37(5): 515-521, 2017 Oct.
Artigo em Inglês | MEDLINE | ID: mdl-28812969

RESUMO

BACKGROUND: Bone morphogenetic proteins (BMPs) are members of the transforming growth factor-ß (TGF-ß) superfamily. Recently, BMP7 has been demonstrated to be produced by salivary glands and contribute to embryonic branching in mice. The BMP7 in saliva is thought to be delivered to the oral cavity and is expected to contact with stratified squamous epithelial cells which line the surface of oral mucosa. In this study, we attempted to investigate the effects of BMP7 on oral epithelial cells. METHODS: The expression of BMP receptors was examined by reverse transcriptase-polymerase chain reaction (RT-PCR). OSCCs were stimulated with human recombinant BMP7 (hrBMP7) and the phosphorylation status of Smad1/5/8 was examined by western blotting. For microarray analysis, Ca9-22 cells were stimulated with 100 ng/mL of hrBMP7 and total RNA was extracted and subjected to real-time PCR. The 5'-untranslated region (5'-UTR) of IL-17 F gene was cloned to pGL4-basic vector and used for luciferase assay. Ca9-22 cells were pre-incubated with DM3189, a specific inhibitor of Smad1/5/8, for inhibition assay. RESULTS: All isoforms of type I and type II BMP receptors were expressed in both Ca9-22 and HSC3 cells and BMP7 stimulation resulted in the phosphorylation of Smad1/5/8 in both cell lines. The microarray analysis revealed the induction of interleukin-17 F (IL-17 F), netrin G2 (NTNG2) and hyaluronan synthase 1 (HAS1). Luciferase assay using the 5'-UTR of the IL-17 F gene revealed transcriptional regulation. Induced IL-17 F production was further confirmed at the protein level by ELISA. Smad1/5/8 inhibitor pretreatment decreased IL-17 F expression levels in the cells.


Assuntos
Proteína Morfogenética Óssea 7/genética , Carcinoma de Células Escamosas/genética , Interleucina-17/genética , Neoplasias Bucais/genética , Proteínas Recombinantes/genética , Proteína Morfogenética Óssea 7/administração & dosagem , Proteína Morfogenética Óssea 7/metabolismo , Carcinoma de Células Escamosas/patologia , Células Epiteliais/metabolismo , Células Epiteliais/patologia , Proteínas Ligadas por GPI/genética , Regulação Neoplásica da Expressão Gênica/efeitos dos fármacos , Humanos , Hialuronan Sintases/genética , Neoplasias Bucais/patologia , Netrinas/genética , Fosforilação , Proteínas Recombinantes/administração & dosagem , Proteínas Recombinantes/metabolismo , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/antagonistas & inibidores , Proteínas Smad/genética , Fator de Crescimento Transformador beta/genética
20.
Drug Des Devel Ther ; 11: 2179-2191, 2017.
Artigo em Inglês | MEDLINE | ID: mdl-28794616

RESUMO

Liver fibrosis is a major pathological feature of chronic liver diseases and there is no effective therapy program at present. Schisandrin B (Sch B) is the major bioactive ingredient of Schisandra chinensis, with antioxidative, anti-inflammatory, antitumor, and hepatoprotective properties. This study aimed to investigate the protective effect and related molecular mechanism of Sch B against carbon tetrachloride (CCl4)-induced liver fibrosis in rats. The in vivo therapeutic effect of Sch B on liver fibrosis induced by CCl4 was examined in rats. In vitro, rat hepatic stellate cells (HSC-T6) were used to assess the effect of Sch B on the activation of HSCs. Sch B effectively attenuated liver damage and progression of liver fibrosis in rats, as evidenced by improved liver function and decreased collagen deposition. The effects of Sch B were associated with attenuating oxidative stress by activating nuclear factor-erythroid 2-related factor 2 (Nrf2)-mediated antioxidant signaling and suppressing HSC activation by inhibiting the transforming growth factor-ß (TGF-ß)/Smad signaling pathway. In an in vitro study, it was shown that Sch B inhibited TGF-ß-induced HSC activation. Finally, Sch B significantly inhibited TGF-ß1-stimulated phosphorylation of Smad and signaling of mitogen-activated protein kinases. This study demonstrates that Sch B prevents the progression of liver fibrosis by the regulation of Nrf2-ARE and TGF-ß/Smad signaling pathways, and indicates that Sch B can be used for the treatment of liver fibrosis.


Assuntos
Lignanas/farmacologia , Cirrose Hepática/tratamento farmacológico , Fator 2 Relacionado a NF-E2/antagonistas & inibidores , Compostos Policíclicos/farmacologia , Transdução de Sinais/efeitos dos fármacos , Proteínas Smad/antagonistas & inibidores , Fator de Crescimento Transformador beta/antagonistas & inibidores , Animais , Tetracloreto de Carbono/antagonistas & inibidores , Ciclo-Octanos/química , Ciclo-Octanos/farmacologia , Lignanas/química , Cirrose Hepática/induzido quimicamente , Cirrose Hepática/metabolismo , Cirrose Hepática/patologia , Masculino , Estrutura Molecular , Fator 2 Relacionado a NF-E2/metabolismo , Compostos Policíclicos/química , Ratos , Ratos Wistar , Proteínas Smad/metabolismo , Fator de Crescimento Transformador beta/metabolismo
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA